18.11.2012 Views

car and pxr increase insig-1 expression - Molecular Pharmacology

car and pxr increase insig-1 expression - Molecular Pharmacology

car and pxr increase insig-1 expression - Molecular Pharmacology

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Molecular</strong> <strong>Pharmacology</strong> Fast Forward. Published on February 21, 2008 as doi:10.1124/mol.107.041012<br />

MOL Manuscript #41012<br />

REGULATORY CROSSTALK BETWEEN DRUG<br />

METABOLISM AND LIPID HOMEOSTASIS: CAR AND PXR<br />

INCREASE INSIG-1 EXPRESSION<br />

Adrian Roth, Renate Looser, Michel Kaufmann, Sharon Blaettler, Franck Rencurel, Wendong<br />

Huang, David D. Moore<br />

<strong>and</strong> Urs A. Meyer<br />

Division of <strong>Pharmacology</strong>/Neurobiology, Biozentrum of the University of Basel, Klingelbergstrasse<br />

50-70, CH-4056 Basel, Switzerl<strong>and</strong> (AR, RL, MK, SB, FR, UAM); Department of Gene Regulation<br />

& Drug Discovery, Beckman Research Institute, City of Hope National Medical Center, 1500 East<br />

Duarte Road, Duarte, CA 91010, USA (WH); Department of <strong>Molecular</strong> <strong>and</strong> Cellular Biology, Baylor<br />

College of Medicine, One Baylor Plaza, Houston, TX 77030, USA (DDM)<br />

Copyright 2008 by the American Society for <strong>Pharmacology</strong> <strong>and</strong> Experimental Therapeutics.<br />

1


MOL Manuscript #41012<br />

Running Title: CAR <strong>and</strong> PXR induce Insig-1<br />

Corresponding Author: Urs A. Meyer<br />

Number of Text Pages : 25<br />

Number of Tables : 1<br />

Number of Figures : 6<br />

Number of References : 31<br />

Division of <strong>Pharmacology</strong>/Neurobiology<br />

Biozentrum of the University of Basel<br />

Klingelbergstrasse 50-70<br />

CH-4056 Basel<br />

Switzerl<strong>and</strong><br />

Phone +41 61 267 22 20<br />

Fax +41 61 267 22 08<br />

Number of words in the Abstract: 247<br />

Number of words in the Introduction: 437<br />

Number of words in the Discussion: 974<br />

Email Urs-A.Meyer@unibas.ch<br />

Nonst<strong>and</strong>ard abbreviations used : CAR, constitutive <strong>and</strong>rostane receptor; CYP, cytochromes P450;<br />

ER, endoplasmatic reticulum, HA, hemagglutinin; PB, Phenobarbital;<br />

PCN, pregnenolone-16α-<strong>car</strong>bonitrile; PXR, pregnane x receptor;<br />

TCPOBOP, 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene; AICAR, 5-<br />

aminoimidazole-4-<strong>car</strong>boxamide riboside<br />

2


ABSTRACT<br />

MOL Manuscript #41012<br />

Activation of PXR (pregnane x receptor) <strong>and</strong> CAR (constitutive <strong>and</strong>rostane receptor) by xenobiotic<br />

inducers of cytochromes P450 is part of a pleiotropic response that includes liver hypertrophy, tumor<br />

promotion, effects on lipid homeostasis <strong>and</strong> energy metabolism. Here we describe an acute response<br />

to CAR <strong>and</strong> PXR activators that is associated with induction of Insig-1, a protein with antilipogenic<br />

properties. We first observed that activation of CAR <strong>and</strong> PXR in mouse liver results in activation of<br />

Insig-1 along with reduced protein levels of the active form of sterol regulatory element binding<br />

protein 1 (Srebp-1). Studies in mice deficient in CAR <strong>and</strong> PXR revealed that the effect on<br />

triglycerides involves these two nuclear receptors. Finally, we identified a functional binding site for<br />

CAR <strong>and</strong> PXR in the Insig-1 gene by in vivo, in vitro <strong>and</strong> in silico genomic analysis. Our experiments<br />

suggest that activation of Insig-1 by drugs leads to reduced levels of active Srebp-1 <strong>and</strong> consequently<br />

to reduced target gene <strong>expression</strong> including the genes responsible for triglyceride synthesis. The<br />

reduction in nuclear Srebp-1 by drugs is not observed when Insig-1 <strong>expression</strong> is repressed by siRNA.<br />

In addition, we observed that Insig-1 is also a target of AMP-activated kinase (AMPK), the hepatic<br />

activity of which is <strong>increase</strong>d by activators of CAR <strong>and</strong> PXR, <strong>and</strong> which is known to cause a<br />

reduction of triglycerides. The fact that drugs which serve as CAR or PXR lig<strong>and</strong>s induce Insig-1<br />

might have clinical consequences <strong>and</strong> explains alterations in lipid levels after drug therapy.<br />

3


MOL Manuscript #41012<br />

Induction of cytochromes P450 (CYPs), other drug-metabolizing enzymes <strong>and</strong> drug transporters by<br />

their own substrates <strong>and</strong> other chemicals is an adaptive response of the liver to prevent accumulation<br />

of toxic xenobiotics <strong>and</strong> endobiotics. Xenosensors that mediate this response are the nuclear receptors<br />

pregnane X receptor (PXR) <strong>and</strong> constitutive active/<strong>and</strong>rostane receptor (CAR; for a review see<br />

(H<strong>and</strong>schin <strong>and</strong> Meyer, 2003)). PXR <strong>and</strong> CAR form heterodimers with the retinoid X receptor (RXR)<br />

<strong>and</strong> bind to specific DNA sequences in the regulatory region of target genes. PXR <strong>and</strong> CAR induce an<br />

overlapping set of genes involved in metabolism <strong>and</strong> transport of drugs (Maglich et al., 2002) but also<br />

genes involved in the regulation of steroids, bile acids, eicosanoids <strong>and</strong> genes involved in cholesterol<br />

<strong>and</strong> bile acid homeostasis (Huang et al., 2003; Staudinger et al., 2001) .<br />

Insig-1 <strong>and</strong> Insig-2 are proteins of endoplasmatic reticulum (ER) membrane <strong>and</strong> play an important<br />

role in the control of triglyceride <strong>and</strong> cholesterol biosynthesis (Yabe et al., 2002; Yang et al., 2002).<br />

The two isoforms bind in a sterol-dependent fashion to another ER membrane protein, sterol<br />

regulatory element binding protein (Srebp) cleavage-activating protein, or Scap, a transport protein<br />

needed for escort <strong>and</strong> subsequent activation of Srebp transcription factors (Hua et al., 1996). When<br />

Insig proteins are activated by sterols, insulin or other stimuli, they retain the Scap-Srebp complex in<br />

the ER membrane thereby preventing Srebp-dependent target gene <strong>expression</strong>. Srebp’s are a group of<br />

basic helix loop helix transcription factors, which activate an array of genes involved in the synthesis<br />

of cholesterol <strong>and</strong> triglycerides. While Srebp-2 is mainly involved in cholesterol biosynthesis, Srebp-<br />

1a <strong>and</strong> Srebp1c mainly activate genes involved in fatty acid <strong>and</strong> triglyceride synthesis (Shimano,<br />

2001).<br />

A decrease in hepatic <strong>and</strong>/or serum lipids, in particular triglycerides, has been observed in rodents<br />

after treatment with inducers of xenobiotic metabolism many years ago (Bjondahl, 1978; Hall et al.,<br />

1990; Venkatesan et al., 1994). More recently, known inducers of human drug metabolism such as the<br />

commonly used anti-retroviral drug efavirenz or the barbiturate phenobarbital (PB) also have been<br />

shown to inhibit lipogenesis (Hadri et al., 2004; Kiyosawa et al., 2004). The molecular mechanism of<br />

the effect of inducers on triglycerides has not been explained.<br />

4


MOL Manuscript #41012<br />

In the present study we show that nuclear receptors CAR <strong>and</strong> PXR transcriptionally activate Insig-1<br />

by binding to an enhancer sequence of the Insig-1 gene. Our results explain the negative effect of<br />

drugs <strong>and</strong> xenobiotics on hepatic lipids in vivo <strong>and</strong> show that CAR <strong>and</strong> PXR not only play a role in<br />

the catabolism of various endogenous <strong>and</strong> exogenous compounds but also directly affect lipogenic<br />

pathways by activating Insig-1. Moreover, as Insig-1 has recently been found to be a possible drug<br />

target for the treatment of diabetes (Nakagawa et al., 2006), this study contributes to the<br />

underst<strong>and</strong>ing of the regulation of this gene <strong>and</strong> possibly to the development of new therapies against<br />

dislipidemia.<br />

5


MATERIALS & METHODS<br />

Animals<br />

MOL Manuscript #41012<br />

C57BL/6 mice were maintained in a 12-hour light/12-hour dark cycle <strong>and</strong> had free access to food <strong>and</strong><br />

drinking water. 9-11 week old male animals received an i.p. injection of either 100 mg/Kg<br />

phenobarbital (PB, Sigma, Buchs, Switzerl<strong>and</strong>), 10 mg/Kg 1,4-Bis[2-(3,5-<br />

dichloropyridyloxy)]benzene (TCPOBOP; Bayer, Leverkusen, FRG) or 40 mg/Kg pregnenolone-16α-<br />

<strong>car</strong>bonitrile (PCN; Sigma, Buchs, Switzerl<strong>and</strong>) in a 5% DMSO-corn oil solution i.p. or with vehicle<br />

10h before dissection. Animals were sacrificed by exposure to CO2, blood was collected by heart<br />

puncture, livers excised <strong>and</strong> snap-frozen in liquid nitrogen <strong>and</strong> stored at -80°C until use.<br />

Analysis of Triglycerides <strong>and</strong> Cholesterol<br />

50-100 mg of liver was used for each preparation. After weight determination, liver samples were put<br />

in an ethanol: ether (3:1, v/v) mixture in Fastprep tubes (Lysing matrix D, Qbiogene, Basel,<br />

Switzerl<strong>and</strong>). Livers were homogenized on the Fastprep instrument for 40s at position 6,5 <strong>and</strong><br />

evaporated to complete dryness on a Speed-vac evaporator. Samples were redissolved in 1ml<br />

isopropanol <strong>and</strong> tissue remnants spun down for 5 minutes at 14000 xg. 600 µl of the supernatant was<br />

mixed with 400 µl of water <strong>and</strong> lipids were determined using the esterase/oxidase kit for cholesterol<br />

determination <strong>and</strong> the L-α-glycerol phosphate oxidase kit for the determination of triglycerides<br />

(Roche Diagnostics, Rotkreuz, Switzerl<strong>and</strong>).<br />

RT PCR Analysis<br />

RNA from cells <strong>and</strong> tissues was isolated using Tri-Reagent (Sigma, Buchs, Switzerl<strong>and</strong>). One<br />

microgram total RNA was reverse-transcribed with MMLV reverse transcriptase (Roche <strong>Molecular</strong><br />

Biochemicals, Rotkreuz, Switzerl<strong>and</strong>). PCR was performed using the TaqMan PCR Core Reagent Kit<br />

(PE Applied Biosystems, Rotkreuz, Switzerl<strong>and</strong>) <strong>and</strong> the transcript level quantitated with an ABI<br />

6


MOL Manuscript #41012<br />

PRISM 7700 Sequence Detection System (PE Applied Biosystems, Rotkreuz, Switzerl<strong>and</strong>) according<br />

to the manufacturer’s protocol. Briefly, relative transcript levels were determined using the relative<br />

quantitation method by measuring the ∆Ct between the gene of interest <strong>and</strong> the internal control<br />

GAPDH. Primers <strong>and</strong> fluorescent probes used in these PCRs are listed in Table 1.<br />

Reporter Gene Assays<br />

Culture <strong>and</strong> transfection of CV-1 cells with Lipofectamine Transfection Reagent (Invitrogen,<br />

Carlsbad, USA) was performed as previously published (H<strong>and</strong>schin et al., 2000). Expression vectors<br />

encoding mouse PXR <strong>and</strong> mouse CAR as well as the beta-galactosidase vector used for signal<br />

normalization have been described (H<strong>and</strong>schin et al., 2002). For construction of hemaglutinin-tagged<br />

vp16 fusion proteins nuclear receptor sequences were amplified from <strong>expression</strong> plasmids <strong>and</strong><br />

subcloned into pcDNA3/vp16-HA (a kind gift from Dr. Dieter Kressler, Biozentrum, University of<br />

Basel, Switzerl<strong>and</strong>). Reporter vectors were based on PGL3-LUC (Promega, Wallisellen, Switzerl<strong>and</strong>).<br />

Genomic DNA from the murine Insig1 enhancer region was amplified using PCR primers <strong>car</strong>rying<br />

restriction sites suitable for direct subcloning into the reporter vector.<br />

Preparation of Primary Hepatocytes<br />

For the preparation of mouse hepatocytes animals were anaesthetized with Ketamine/Xylazine (Sigma,<br />

Buchs, Switzerl<strong>and</strong>). The portal vein was canulated <strong>and</strong> perfused with HEPES-EGTA (pH 7.4) for 5<br />

minutes <strong>and</strong> then with collagenase (type 2, Worthington, Lakewood NJ, USA) for 6 minutes. The<br />

livers were excised, cells were filtered through a nylon mesh <strong>and</strong> centrifuged at 50 xg at 4°C for 5<br />

minutes three times. After determination of viability, cells were plated at a density of 400’000<br />

cells/well (12 well plate) <strong>and</strong> were allowed to attach for 2 hours in William’s E medium without<br />

phenol red (Invitrogen, Basel, Switzerl<strong>and</strong>), 10 % FCS, 4µg/ml insulin, 200µM glutamine <strong>and</strong> 1%<br />

penicillin/streptomycin (50 IU/ml) on collagen-coated dishes. Induction experiments were performed<br />

7


MOL Manuscript #41012<br />

in the same medium without FCS <strong>and</strong> with reduced insulin (2 µg/ml) but with the addition of 1 µM<br />

hydrocortisone.<br />

Primary human hepatocytes in suspension were allowed to attach on collagen-coated plates in DMEM<br />

supplemented with 10% FBS, 1% penicillin/streptomycin (50 IU/ml) <strong>and</strong> 1 µM dexamethasone<br />

overnight before start of the experiments. For induction cells were cultured in DMEM without serum<br />

but supplemented with insulin-transferrine-selenium mixture (Sigma, Buchs, Switzerl<strong>and</strong>) <strong>and</strong> 1µM<br />

hydrocortisone<br />

Production of recombinant adenovirus particles<br />

Expression cassettes of interest were PCR-amplified using vector-specific primers with attB1/attB2-<br />

Gateway extensions for subsequent cloning into pDONR221 (Invitrogen, Carlsbad, CA, USA). For<br />

pcDNA3 constructs (Vp16-PXR, Vp16-CAR) the following primers were used:<br />

TTAGGGTTAGGCGTTTTGCGC (Fwd), TCAGAAGCCATAGAGCCCAC (Rev). Entry clones<br />

<strong>car</strong>rying the PCR products were used for cloning into pAd-DEST (Invitrogen, Carlsbad, CA, USA).<br />

PacI-digested plasmids were transfected into HEK293A cells <strong>and</strong> adenovirus particles produced <strong>and</strong><br />

processed according to the manufacturer’s recommendations (Invitrogen, Carlsbad, CA, USA).<br />

Functionality of PXR- <strong>and</strong> CAR-expressing adenoviruses was assessed in reporter gene assays in CV-<br />

1 cells using PXR- <strong>and</strong> CAR-responsive reporter vectors. We also tested the recombinant proteins in<br />

mouse hepatocytes by measuring mRNA <strong>expression</strong> of the target genes Cyp3a11 <strong>and</strong> Cyp2b10,<br />

respectively (data not shown).The adenovirus particles encoding recombinant forms of AMPK have<br />

been described (20)<br />

Immunoblotting, Gel-Mobility-Shift Assay & Chromatin Immunoprecipitation<br />

For Western blot analysis of SREBP1, liver proteins were extracted from 100-200 mg of frozen tissue<br />

in 1 ml ice-cold buffer (50 mM Tris-HCl pH 7.4, 100mM KCl, 1mM EDTA pH 8.0, 10mM beta-<br />

Mercaptoethanol, 5mM DTT, 0.1% (v/v) Triton X-100, 0.1% (v/v) NP 40, 1 tablet/50ml buffer<br />

8


MOL Manuscript #41012<br />

Protease Inhibitor Cocktail (Roche Diagnostics, Rotkreuz, Switzerl<strong>and</strong>)) in a 5 ml polystyrene tube<br />

using a Polytron Rotor-Stator Homogenizer. The homogenate was centrifuged for 30 min at 10000<br />

rpm at 4°C <strong>and</strong> 50 µg of protein was loaded on a 10% SDS-gel. SREBP1 isoforms were detected<br />

using mouse anti-SREBP1 monoclonal antibody (Anti-SREBP1 monoclonal #557036, Clone IgG-<br />

2A4BD, Pharmingen, Allschwil, Switzerl<strong>and</strong>).<br />

Transcription factors were synthesized in vitro by using the TNT T7 Quick Coupled<br />

Transcription/Translation System (Promega, Wallisellen, Switzerl<strong>and</strong>) according to the<br />

manufacturer's instructions. Probes were labeled with the Klenow fragment of DNA polymerase in the<br />

presence of radiolabeled [ -32P]ATP, <strong>and</strong> the probe was purified over a Biospin 6 chromatography<br />

column. A volume of labelled oligonucleotide corresponding to 100,000 cpm was used for each<br />

reaction in 10 mM Tris HCl, pH 8.0/40 mM KCl/0.05% Nonidet P-40/6% (vol/vol) glycerol/1 mM<br />

DTT containing 0.2 µg of poly(dI-dC) poly(dI-dC) <strong>and</strong> 2.5 µl of the in vitro synthesized proteins as<br />

described previously (H<strong>and</strong>schin et al.). The mix was incubated for 20 min at room temperature <strong>and</strong><br />

subsequently electrophoresed on a 6% polyacrylamide gel in 0.5× Tris/borate/EDTA buffer (1x TBE<br />

buffer is composed of 0.9 M Tris-Borate, 0.002 M EDTA, pH 8.3) followed by autoradiography at<br />

70°C. Oligos used for EMSAs were obtained as follows: For the mouse Insig-1 DR4 the following<br />

oligonucleotide were annealed <strong>and</strong> labelled using polynucleotide kinase:<br />

CCTGAGGGTCAACAGAGGACACCTAG (Fwd) <strong>and</strong> CTAGGTGTCCTCTGTTGACCCTCAGG<br />

(Rev).<br />

Chromatin Immunoprecipitation was performed using the EZ-Chip kit from Upstate<br />

(Charlottesville, USA). Primary mouse hepatocytes were infected with adenoviral particles encoding<br />

HA-tagged vp16-mouse CAR or mouse PXR, respectively. After 24 hours cells were harvested <strong>and</strong><br />

samples processed according to the manufacturers recommendations. For immunoprecipitation<br />

hemagglutinin (HA) antibody (monoclonal HA.11 clone 16B12 mouse IgG1 MMS-101P) from<br />

Covance (Princeton, USA)was used.<br />

9


MOL Manuscript #41012<br />

Targeting of Insig-1 in primary mouse hepatocytes by siRNA<br />

For the transfection of primary mouse hepatocytes, Dharmafect1 transfection reagent (#T2001-01,<br />

Dharmacon, Chicago, USA) was used. 100nM siRNAs (siGenome SmartPool mINSIG1 #M-060068-<br />

00; siControl Nontargeting siRNAPool#2 #D-001206-14-05; Dharmacon, Chicago, USA) <strong>and</strong> 14µl<br />

Dharmafect were used according to the manufacturer’s instructions. 6h after transfection medium was<br />

removed <strong>and</strong> replaced with fresh medium without serum. 24h later, medium was replace by medium<br />

containing 500µM phenobarbital <strong>and</strong> mRNAs <strong>and</strong> Srebp-1 protein analyzed after 24h as described.<br />

10


RESULTS<br />

MOL Manuscript #41012<br />

Mice were injected with drugs known to activate nuclear receptors PXR <strong>and</strong> CAR, namely<br />

phenobarbital (PB, activator of both PXR <strong>and</strong> CAR), pregnenolone-16α-<strong>car</strong>bonitrile (PCN, activator<br />

of PXR) <strong>and</strong> 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP, activator of CAR; Fig. 1).<br />

After 10 hours of exposure, liver samples were analyzed for hepatic triglycerides <strong>and</strong> cholesterol (Fig.<br />

1, top panel). All three drugs caused a substantial drop in triglycerides, whereas cholesterol levels<br />

were less affected. PB <strong>and</strong> TCPOBOP at the doses applied resulted in a 49% or 67% decrease in<br />

triglycerides, respectively, <strong>and</strong> a 28% or 33% decrease in cholesterol. Serum analysis of PB-treated<br />

animals revealed no significant change in triglycerides or cholesterol at 10 hours (Fig. 1, top panel,<br />

right). RT-PCR analysis of these livers showed marked induction of Insig-1 mRNA (Fig. 1, middle<br />

panel). The mRNA levels of Insig-2 were not significantly induced by drug treatment (data not<br />

shown) <strong>and</strong> mRNA levels of Srebp genes remained unaffected by drug treatment as well. Accordingly,<br />

Hmg-CoA reductase (Hmgcr), a target gene of Srebp-2 (Horton et al., 1998) was unchanged while<br />

Stearoyl-CoA desaturase 1 (Scd-1), which is a target gene of Srebp-1 (Shimano et al., 1999), was<br />

reduced after drug treatment. Again, TCPOBOP showed strongest effects (Fig. 1, middle panel).<br />

Whether the reduction in hepatic triglycerides was due to reduced nuclear <strong>expression</strong> of Srebp-1 was<br />

tested by immunoblotting of liver protein extracts from drug-treated animals using an antibody which<br />

can discriminate between the inactive precursor form of Srebp1 <strong>and</strong> the activated nuclear (mature)<br />

form of Srebp-1 (Fig. 2A). The graph shows a reduction in nuclear content of Srebp-1 in drug-treated<br />

mice with strongest effects by PB <strong>and</strong> TCPOBOP. A time course experiment in primary mouse<br />

hepatocytes revealed that the time-dependent induction profile of Insig-1 mRNA paralleled the one of<br />

the classic CAR- <strong>and</strong> PXR-inducible gene Cyp2b10 (Fig. 2B).<br />

To define the role for PXR <strong>and</strong> CAR in the activation of Insig1 <strong>and</strong> the subsequent drop in hepatic<br />

triglycerides we applied PB to mice deficient in these two receptors (Zhang et al., 2004), Fig. 3). Fig.<br />

3 shows that two typical target genes of PXR <strong>and</strong> CAR, Cyp2b10 <strong>and</strong> Cyp3a11, expectedly were not<br />

11


MOL Manuscript #41012<br />

inducible by PB in PXR/CAR null mice. Blunted induction of Insig-1 mRNA in these animals after<br />

PB treatment as well as an unchanged triglyceride profile compared to wild type animals was<br />

observed (Fig. 3, lower panels). These data support a PXR/CAR-dependent mechanism for the<br />

triglyceride-lowering effect of inducer compounds.<br />

We therefore designed experiments to identify functional binding sites for the PXR <strong>and</strong> CAR in the<br />

regulatory region of Insig-1. Two 3kb fragments of genomic DNA from the 5’-flanking region of the<br />

Insig1 gene were cloned into a luciferase reporter vector <strong>and</strong> activity was assessed in transactivation<br />

assays in CV-1 cells (Fig. 4A). The first DNA element spanned the transcriptional start site including<br />

the proximal promoter of Insig1 to -3044 bp <strong>and</strong> showed no activation after drug treatment. The<br />

second large DNA stretch was overlapping with the first one <strong>and</strong> ended at –6252bp. There was slight<br />

(as compared to empty control luciferase vector) activation of reporter gene transcription after drug<br />

treatment. This DNA element was cut into smaller pieces <strong>and</strong> activity assessed until a 760 bp<br />

fragment revealed a robust response to PXR <strong>and</strong> CAR. Within this fragment a DR-4 type drug<br />

response element was identified using the Nubiscan algorithm (Podvinec et al., 2002). This element<br />

responded well to PXR <strong>and</strong> CAR <strong>and</strong> specificity was assessed using a mutated version of the DR-4<br />

element, which resulted in a decreased response to drugs (Fig. 4A). The same DR-4 element was used<br />

in electromobility shift assays together with in vitro translated PXR, CAR <strong>and</strong> their heterodimeric<br />

partner, RXR (Fig. 4B). A strong b<strong>and</strong> was observed when CAR <strong>and</strong> RXR were incubated with the<br />

oligonucleotide <strong>car</strong>rying the DR-4 element <strong>and</strong> less intensive binding appeared when PXR was used.<br />

Both nuclear receptor complexes were super-shifted by co-incubation with an anti-RXR antibody.<br />

Functionality of this element was tested in vivo by chromatin immunoprecipitation in primary mouse<br />

hepatocytes (Fig. 4C). Cells were infected with adenovirus particles expressing HA-tagged versions<br />

of both CAR <strong>and</strong> PXR. The amplified PCR product corresponds to the 157bp region in the murine<br />

Insig-1 promoter where the designated DR-4 element is located. In Fig 5B a reporter gene analysis<br />

using the Insig-1 DR-4 with inducers of mouse PXR <strong>and</strong> CAR, respectively, was performed <strong>and</strong><br />

12


MOL Manuscript #41012<br />

revealed a 4-fold induction after PCN-treatment <strong>and</strong> a 7.2-fold induction after TCBOBOP-treatment<br />

(Fig. 5A).<br />

A role for Insig-1 in mediating the repressive effects of inducer drugs was established by siRNA-<br />

mediated inhibition of Insig-1 <strong>expression</strong> <strong>and</strong> concurrent treatment with PB (Fig. 5B). Primary mouse<br />

hepatocytes were transfected with either unspecific control siRNAS or siRNA targeting Insig-1. After<br />

48h, mRNA analysis revealed markedly reduced Insig-1 <strong>expression</strong> (Fig. 5B, left panel). Immunoblot<br />

analysis of Srebp-1 protein levels in siRNA transfected cultures treated with or without PB was<br />

performed (Fig. 5B, right panel). The results reveal reduced nuclear <strong>expression</strong> of Srebp-1 in cells<br />

transfected with control siRNA <strong>and</strong> treated with PB. In cells where Insig-1 <strong>expression</strong> was reduced by<br />

siRNA, Srebp-1 protein levels in the nucleus remained unaffected (Fig. 5B, right panel).<br />

To reveal a potential role of this mechanism in humans we tested the inducibility of human Insig1 by<br />

PB in primary human hepatocytes (Fig. 6A). Treatment for 50 hours with PB resulted in a significant<br />

induction of Insig-1 in cultures of two different donors <strong>and</strong> this was paralleled by a reduction in<br />

Srebp1c <strong>expression</strong>. Induction of CYP2B6 <strong>and</strong> CYP3A4 served as positive controls.<br />

As recently reported, induction of drug metabolizing enzymes requires activation of AMP-activated<br />

kinase (Rencurel et al., 2006; Rencurel et al., 2005). We therefore wanted to test whether this kinase,<br />

alone or in combination with drug, can regulate transcription of Insig1 (Fig. 6B). Primary human<br />

hepatocytes were infected with control virus (expressing beta-galactosidase) or different versions of<br />

AMPK: a dominant negative construct (kinase dead, KD), the alpha-1 or the alpha-2 subunit of<br />

AMPK. While the dominant negative version repressed <strong>expression</strong> of Insig1, both subunits induced<br />

Insig1 with stronger effects seen using the alpha-1 subunit. Furthermore, exposure to PB enhanced<br />

these effects (Fig. 6B).<br />

13


DISCUSSION<br />

MOL Manuscript #41012<br />

The experiments described here reveal a novel molecular mechanism by which drugs that induce<br />

drug-metabolizing enzymes <strong>and</strong> drug transporters can acutely regulate hepatic triglyceride levels.<br />

While several CYPs <strong>and</strong> other enzymes involved in metabolism <strong>and</strong> transport of xenochemicals have<br />

been known to be targets of nuclear receptors CAR <strong>and</strong> PXR, the upregulation of Insig-1 by the same<br />

receptors after drug treatment is new <strong>and</strong> adds a potentially clinically important aspect to the present<br />

underst<strong>and</strong>ing on how the liver reacts to accumulating lipophilic compounds such as PB. This report<br />

shows that the drug metabolizing process also includes direct regulation of hepatic lipid biosynthesis<br />

by induction of an important regulatory protein as is Insig-1. We present evidence for a functional<br />

DR-4 binding site for CAR <strong>and</strong> PXR in the upstream promoter region of Insig-1 (Fig. 4). Binding of<br />

the xenobiotic receptors to this DR-4 site can account for the induction of Insig-1, which results in the<br />

reduced <strong>expression</strong> of the activated nuclear form of Srebp-1 <strong>and</strong> the substantial reduction in hepatic<br />

triglycerides after only 10 hours of treatment (Fig. 1,2). The fact that Insig-1 has been observed in<br />

<strong>expression</strong> studies to be induced early after treatment with the CAR lig<strong>and</strong> TCPOBOP (Locker et al.,<br />

2003) <strong>and</strong> that over<strong>expression</strong> of Insig-1 in livers of mice has been shown to cause a drop in<br />

triglyceride levels with smaller effects on cholesterol (Engelking et al., 2004; Takaishi et al., 2004)<br />

made Insig-1 an interesting c<strong>and</strong>idate gene for transcriptional regulation by CAR <strong>and</strong> PXR. We first<br />

established that in CAR/PXR double knockout mice PB had lost its effect on triglycerides <strong>and</strong> there<br />

was no induction of Insig-1 (Fig. 3). This strongly supported the idea of a CAR/PXR-mediated<br />

transcriptional activation of the Insig-1 gene. Moreover, the induction of Insig1 mRNA appeared early<br />

after addition of drug (Fig. 2B) making a rapid decrease in activated nuckear form of Srebp-1 protein<br />

levels a plausible scenario (Fig. 2A). The more pronounced effects on triglycerides seen with the CAR<br />

activators TCPOBOP <strong>and</strong> PB compared to the PXR lig<strong>and</strong> PCN is in line with the more pronounced<br />

activation of the Insig1 promoter by CAR (Fig. 4A, 5B), with the higher affinity of CAR to bind to<br />

the DR-4 element (Fig. 4B) as well as with the stronger enrichment in CAR-immunoprecipitated<br />

14


MOL Manuscript #41012<br />

samples of this fragment (Fig. 4C). A role for Insig-1 in mediating these effects was established in<br />

mouse hepatocytes with siRNA-reduced Insig-1 <strong>expression</strong> (Fig. 5B). While the repressive effect of<br />

PB on nuclear protein levels of Srebp-1 was evident, in cells with repressed Insig-1 <strong>expression</strong> this<br />

effect was not detectable, strongly supporting the concept that PXR/CAR-activated Insig-1 is<br />

responsible for reduced Srebp-1 <strong>and</strong> thereby for lowered hepatic triglycerides. Moreover, the fact that<br />

these effects could be reproduced in human hepatocytes supports the concept of a general mechanism<br />

by which drugs affect hepatic lipid biosynthesis (Fig. 6A). Insig- 2, the other member of the Insig<br />

family of Srebp-regulating genes was not affected by PXR/CAR inducers <strong>and</strong> a potential role of this<br />

gene in linking drug treatment to reduced triglyceride levels was not pursued. It cannot be ruled out of<br />

course, that under different conditions, this gene may also play a role in mediating lipid synthesis due<br />

to a xenobiotic challenge.<br />

An effect of PXR on lipogenic genes has recently been described by Nakamura et al. (Nakamura et<br />

al., 2007). In strong support of our data they observed downregulation of lipogenic genes by the PXR-<br />

specific activator PCN <strong>and</strong> this effect was not seen in PXR -/- mice. Interestingely, Nakamura <strong>and</strong><br />

colleagues furthermore observed an induction of Scd-1 in PCN-treated animals that were fasted for<br />

24h. This suggests an additional level of regulation of lipogenesis by xenobiotics in the fasted state.<br />

Another new finding of our study is the role of AMPK in the induction of Insig1 (Fig. 6B). AMPK is<br />

considered a metabolic master-switch sensing cellular energy levels <strong>and</strong> regulating glucose transport<br />

<strong>and</strong> gluconeogenesis. It is activated in response to metabolic stress signals that deplete cellular ATP<br />

<strong>and</strong> stimulate fatty acid oxidation (Kahn et al., 2005). It was recently shown that CAR-dependent<br />

induction of CYP2B by PB requires activation of AMPK (Rencurel et al., 2006). Blattler et al.<br />

(Blattler et al., 2007) demonstrated that PB interferes with mitochondrial function <strong>and</strong> activates the<br />

AMPK upstream kinase LKB1 which then mediates the activation cascade of AMPK to CAR.<br />

Interestingly, AMPK, either via activation by AICAR (5-aminoimidazole-4-<strong>car</strong>boxamide riboside) or<br />

via adenoviral over<strong>expression</strong> of its catalytic subunit, also has been shown to reduce Srebp-1c<br />

<strong>expression</strong> (Foretz et al., 2005; Zhou et al., 2001). As these observations lacked a mechanistic<br />

15


MOL Manuscript #41012<br />

explanation, the activation of Insig-1 by AMPK shown here may indicate a signaling pathway leading<br />

to repression of Srebp-1c. In line with observations by Rencurel et al. (2006), AMPK alone seems<br />

capable of regulating <strong>expression</strong> of CAR/PXR target genes <strong>and</strong> addition of a nuclear receptor<br />

activator leads to a synergistic effect on gene transcription. However, the detailed interplay between<br />

PB, nuclear receptors <strong>and</strong> AMPK in the induction of Insig-1 clearly requires further investigation.<br />

Also, the data presented here account for the immediate physiologic response of the liver to a<br />

xenobiotic challenge. Chronic treatment with drugs leading to constant activation of PXR <strong>and</strong>/or CAR<br />

may lead to diverse adaptive gene-regulations to maintain lipid homeostasis (Kiyosawa et al., 2004;<br />

Zhou et al., 2006).<br />

In conclusion, the results of our experiments suggest that the signaling pathways involved in<br />

mediating the effect of xenobiotics on detoxification also induce Insig-1, a gene regulating lipid<br />

biosynthesis <strong>and</strong> that this is associated with an acute lowering of triglyceride levels in the liver. As<br />

Insig-1 has recently been suggested as a possible drug target for the treatment of dislipidemic diseases<br />

including diabetes (Nakagawa et al., 2006) the observation that CAR <strong>and</strong> PXR lig<strong>and</strong>s or activators<br />

induce Insig-1 may have clinical consequences <strong>and</strong> explains the reported alterations in lipid levels<br />

after drug therapy.<br />

16


ACKNOWLEDGEMENTS<br />

MOL Manuscript #41012<br />

The authors would like to thank Michael Podvinec (Institute of Bioinformatics, University of Basel,<br />

Switzerl<strong>and</strong>) for assistance in in silico sequence analysis <strong>and</strong> Frederic Delobel (Hoffmann-La Roche<br />

Ltd, Basel, Switzerl<strong>and</strong>) for experimental support with primary mouse hepatocytes.<br />

17


REFERENCES<br />

Bjondahl K (1978) A study on cerium-induced liver injury in rats after pretreatment with<br />

spironolactone, phenobarbital, pregnenolone-16 alpha-<strong>car</strong>bonitrile <strong>and</strong> nafenopin.<br />

Arzneimittelforschung 28:817-9.<br />

Blattler SM, Rencurel F, Kaufmann MR <strong>and</strong> Meyer UA (2007) In the regulation of<br />

cytochrome P450 genes, phenobarbital targets LKB1 for necessary activation of<br />

AMP-activated protein kinase. Proc Natl Acad Sci U S A 104:1045-50.<br />

Engelking LJ, Kuriyama H, Hammer RE, Horton JD, Brown MS, Goldstein JL <strong>and</strong> Liang G<br />

(2004) Over<strong>expression</strong> of Insig-1 in the livers of transgenic mice inhibits SREBP<br />

processing <strong>and</strong> reduces insulin-stimulated lipogenesis. J Clin Invest 113:1168-75.<br />

Foretz M, Ancellin N, Andreelli F, Saintillan Y, Grondin P, Kahn A, Thorens B, Vaulont S<br />

<strong>and</strong> Viollet B (2005) Short-term over<strong>expression</strong> of a constitutively active form of<br />

AMP-activated protein kinase in the liver leads to mild hypoglycemia <strong>and</strong> fatty liver.<br />

Diabetes 54:1331-9.<br />

Hadri KE, Glorian M, Monsempes C, Dieudonne MN, Pecquery R, Giudicelli Y, Andreani M,<br />

Dugail I <strong>and</strong> Feve B (2004) In vitro suppression of the lipogenic pathway by the<br />

nonnucleoside reverse transcriptase inhibitor efavirenz in 3T3 <strong>and</strong> human<br />

preadipocytes or adipocytes. J Biol Chem 279:15130-41.<br />

Hall IH, Patrick MA <strong>and</strong> Maguire JH (1990) Hypolipidemic activity in rodents of<br />

phenobarbital <strong>and</strong> related derivatives. Arch Pharm (Weinheim) 323:579-86.<br />

H<strong>and</strong>schin C <strong>and</strong> Meyer UA (2003) Induction of drug metabolism: the role of nuclear<br />

receptors. Pharmacol Rev 55:649-73.<br />

H<strong>and</strong>schin C, Podvinec M, Amherd R, Looser R, Ourlin JC <strong>and</strong> Meyer UA (2002)<br />

Cholesterol <strong>and</strong> bile acids regulate xenosensor signaling in drug-mediated induction<br />

of cytochromes P450. J Biol Chem 277:29561-7.<br />

H<strong>and</strong>schin C, Podvinec M <strong>and</strong> Meyer UA (2000) CXR, a chicken xenobiotic-sensing orphan<br />

nuclear receptor, is related to both mammalian pregnane X receptor (PXR) <strong>and</strong><br />

constitutive <strong>and</strong>rostane receptor (CAR). Proc Natl Acad Sci U S A 97:10769-74.<br />

Horton JD, Shimomura I, Brown MS, Hammer RE, Goldstein JL <strong>and</strong> Shimano H (1998)<br />

Activation of cholesterol synthesis in preference to fatty acid synthesis in liver <strong>and</strong><br />

adipose tissue of transgenic mice overproducing sterol regulatory element-binding<br />

protein-2. J Clin Invest 101:2331-9.<br />

Hua X, Sakai J, Brown MS <strong>and</strong> Goldstein JL (1996) Regulated cleavage of sterol regulatory<br />

element binding proteins requires sequences on both sides of the endoplasmic<br />

reticulum membrane. J Biol Chem 271:10379-84.<br />

Huang W, Zhang J, Chua SS, Qatanani M, Han Y, Granata R <strong>and</strong> Moore DD (2003)<br />

Induction of bilirubin clearance by the constitutive <strong>and</strong>rostane receptor (CAR). Proc<br />

Natl Acad Sci U S A 100:4156-61.<br />

Kahn BB, Alquier T, Carling D <strong>and</strong> Hardie DG (2005) AMP-activated protein kinase: ancient<br />

energy gauge provides clues to modern underst<strong>and</strong>ing of metabolism. Cell Metab<br />

1:15-25.<br />

Kiyosawa N, Tanaka K, Hirao J, Ito K, Niino N, Sakuma K, Kanbori M, Yamoto T, Manabe<br />

S <strong>and</strong> Matsunuma N (2004) <strong>Molecular</strong> mechanism investigation of phenobarbitalinduced<br />

serum cholesterol elevation in rat livers by microarray analysis. Arch Toxicol<br />

78:435-52.


MOL Manuscript #41012<br />

Locker J, Tian J, Carver R, Concas D, Cossu C, Ledda-Columbano GM <strong>and</strong> Columbano A<br />

(2003) A common set of immediate-early response genes in liver regeneration <strong>and</strong><br />

hyperplasia. Hepatology 38:314-25.<br />

Maglich JM, Stoltz CM, Goodwin B, Hawkins-Brown D, Moore JT <strong>and</strong> Kliewer SA (2002)<br />

Nuclear pregnane x receptor <strong>and</strong> constitutive <strong>and</strong>rostane receptor regulate overlapping<br />

but distinct sets of genes involved in xenobiotic detoxification. Mol Pharmacol<br />

62:638-46.<br />

Nakagawa Y, Shimano H, Yoshikawa T, Ide T, Tamura M, Furusawa M, Yamamoto T, Inoue<br />

N, Matsuzaka T, Takahashi A, Hasty AH, Suzuki H, Sone H, Toyoshima H, Yahagi N<br />

<strong>and</strong> Yamada N (2006) TFE3 transcriptionally activates hepatic IRS-2, participates in<br />

insulin signaling <strong>and</strong> ameliorates diabetes. Nat Med 12:107-13.<br />

Nakamura K, Moore R, Negishi M <strong>and</strong> Sueyoshi T (2007) Nuclear Pregnane X Receptor<br />

Cross-talk with FoxA2 to Mediate Drug-induced Regulation of Lipid Metabolism in<br />

Fasting Mouse Liver. J Biol Chem 282:9768-76.<br />

Podvinec M, Kaufmann MR, H<strong>and</strong>schin C <strong>and</strong> Meyer UA (2002) NUBIScan, an in Silico<br />

Approach for Prediction of Nuclear Receptor Response Elements. Mol Endocrinol<br />

16:1269-79.<br />

Rencurel F, Foretz M, Kaufmann MR, Stroka D, Looser R, Leclerc I, da Silva Xavier G,<br />

Rutter GA, Viollet B <strong>and</strong> Meyer UA (2006) Stimulation of AMP-activated protein<br />

kinase is essential for the induction of drug metabolizing enzymes by phenobarbital in<br />

human <strong>and</strong> mouse liver. Mol Pharmacol 70:1925-34.<br />

Rencurel F, Stenhouse A, Hawley SA, Friedberg T, Hardie DG, Sutherl<strong>and</strong> C <strong>and</strong> Wolf CR<br />

(2005) AMP-activated protein kinase mediates phenobarbital induction of CYP2B<br />

gene <strong>expression</strong> in hepatocytes <strong>and</strong> a newly derived human hepatoma cell line. J Biol<br />

Chem 280:4367-73.<br />

Shimano H (2001) Sterol regulatory element-binding proteins (SREBPs): transcriptional<br />

regulators of lipid synthetic genes. Prog Lipid Res 40:439-52.<br />

Shimano H, Yahagi N, Amemiya-Kudo M, Hasty AH, Osuga J, Tamura Y, Shionoiri F,<br />

Iizuka Y, Ohashi K, Harada K, Gotoda T, Ishibashi S <strong>and</strong> Yamada N (1999) Sterol<br />

regulatory element-binding protein-1 as a key transcription factor for nutritional<br />

induction of lipogenic enzyme genes. J Biol Chem 274:35832-9.<br />

Staudinger JL, Goodwin B, Jones SA, Hawkins-Brown D, MacKenzie KI, LaTour A, Liu Y,<br />

Klaassen CD, Brown KK, Reinhard J, Willson TM, Koller BH <strong>and</strong> Kliewer SA<br />

(2001) The nuclear receptor PXR is a lithocholic acid sensor that protects against liver<br />

toxicity. Proc Natl Acad Sci U S A 98:3369-74.<br />

Takaishi K, Duplomb L, Wang MY, Li J <strong>and</strong> Unger RH (2004) Hepatic <strong>insig</strong>-1 or -2<br />

over<strong>expression</strong> reduces lipogenesis in obese Zucker diabetic fatty rats <strong>and</strong> in<br />

fasted/refed normal rats. Proc Natl Acad Sci U S A 101:7106-11.<br />

Venkatesan N, Davidson MB, Simsolo RB <strong>and</strong> Kern PA (1994) Phenobarbital treatment<br />

enhances insulin-mediated glucose metabolism <strong>and</strong> improves lipid metabolism in the<br />

diabetic rat. Metabolism 43:348-56.<br />

Yabe D, Brown MS <strong>and</strong> Goldstein JL (2002) Insig-2, a second endoplasmic reticulum protein<br />

that binds SCAP <strong>and</strong> blocks export of sterol regulatory element-binding proteins.<br />

Proc Natl Acad Sci U S A 99:12753-8.<br />

Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, Goldstein JL <strong>and</strong><br />

Brown MS (2002) Crucial step in cholesterol homeostasis: sterols promote binding of<br />

19


MOL Manuscript #41012<br />

SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER.<br />

Cell 110:489-500.<br />

Zhang J, Huang W, Qatanani M, Evans RM <strong>and</strong> Moore DD (2004) The constitutive<br />

<strong>and</strong>rostane receptor <strong>and</strong> pregnane X receptor function coordinately to prevent bile<br />

acid-induced hepatotoxicity. J Biol Chem 279:49517-22.<br />

Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T,<br />

Fujii N, Musi N, Hirshman MF, Goodyear LJ <strong>and</strong> Moller DE (2001) Role of AMPactivated<br />

protein kinase in mechanism of metformin action. J Clin Invest 108:1167-74.<br />

Zhou J, Zhai Y, Mu Y, Gong H, Uppal H, Toma D, Ren S, Evans RM <strong>and</strong> Xie W (2006) A<br />

novel pregnane X receptor-mediated <strong>and</strong> sterol regulatory element-binding proteinindependent<br />

lipogenic pathway. J Biol Chem 281:15013-20.<br />

20


FOOTNOTES<br />

MOL Manuscript #41012<br />

This work was supported by grants of the Swiss National Science Foundation <strong>and</strong> the STEROLTALK<br />

project (EC contract No. LSHG-CT-2005-512096) under the 6 th framework program.<br />

21


LEGENDS TO FIGURES<br />

Fig. 1<br />

MOL Manuscript #41012<br />

Lipid <strong>and</strong> gene <strong>expression</strong> changes after drug application in mouse liver. PB, TCPOBOP <strong>and</strong><br />

PCN were i.p.-injected in mice <strong>and</strong> livers or serum samples analyzed after 10h. Upper panel,<br />

triglyceride (TG) <strong>and</strong> total cholesterol (CHOL) analysis in liver homogenates <strong>and</strong> blood samples.<br />

Middle <strong>and</strong> lower panel, RT-PCR analysis of RNAs from mouse livers. Bars indicate mean values<br />

from 6 animals per treatment group <strong>and</strong> st<strong>and</strong>ard deviations therefrom. *p


Fig. 4<br />

MOL Manuscript #41012<br />

Analysis of the Insig1 promoter for drug response elements. A, Reporter gene assays using<br />

different genomic DNA sequences from the Insig1 promoter cloned into tk-Luc vector. At the bottom<br />

of the Fig. a Luc vector with a mutated Insig1 DR-4 was used.. Plasmids were cotransfected together<br />

with mouse CAR or PXR into CV-1 cells <strong>and</strong> luciferase activities determined 16h after addition of<br />

1µM TCPOBOP <strong>and</strong> 10µM PCN, respectively. Cell lysates were analyzed for luciferase <strong>expression</strong><br />

<strong>and</strong> fold activation of reporter fragment calculated relative to empty luciferase vector treated with<br />

drugs is indicated. The sequence of the identified DR-4 type drug response element as well as the<br />

mutated form used in the reporter assay is shown in a separate box. B, Gel shift assay using in vitro<br />

translated mouse CAR, PXR <strong>and</strong> RXR <strong>and</strong> a radiolabelled oligonucleotide encoding the putative<br />

Insig1 DR-4 element. The experimental conditions for each lane are indicated in the panel above the<br />

picture: “+” indicates that the corresponding receptor was added, while “-“ indicates that no receptor<br />

was added , * position of shifted nuclear receptor heterodimer, ** position of anti-RXR supershifted<br />

nuclear receptor heterodimer. C, Association of CAR <strong>and</strong> PXR with the Insig1 DR-4 analyzed by<br />

chromatin immunoprecipitation in primary mouse hepatocytes using adenovirus encoding HA-tagged<br />

CAR <strong>and</strong> PXR, respectively. Lower panel shows a control experiment using the same lysates on the<br />

GAPDH promoter.<br />

<strong>and</strong>rostanol (AND), 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) <strong>and</strong> pregnenolone-16α-<br />

<strong>car</strong>bonitrile (PCN)<br />

Fig. 5<br />

Induction of Insig1 DR-4 reporter gene by PXR <strong>and</strong> CAR inducers <strong>and</strong> lack of effect of<br />

Phenobarbital on SREBP-1 proteins when Insig-1 <strong>expression</strong> is suppressed by RNAi. A, Effect<br />

of PXR, CAR <strong>and</strong> their lig<strong>and</strong>s/activators on the Insig-1 DR-4 reporter gene. PXR was activated by<br />

over night treatment with 10µM pregnenolone-16α-<strong>car</strong>bonitrile (PCN). Constitutive active CAR was<br />

23


MOL Manuscript #41012<br />

repressed by the addition of 1µM <strong>and</strong>rostanol <strong>and</strong> activated by addition of 10µM 1,4-Bis[2-(3,5-<br />

dichloropyridyloxy)]benzene (TCPOBOP). Fold activations are calculated as relaitve reporter gene<br />

levels over non-drug-treated cells. B, primary mouse hepatocytes were transfected with either control<br />

siRNA or siRNA against Insig-1. mRNA analysis of Insig-1-siRNA effect after 48h is shown in the<br />

left panel, effect of PB treatment on Srebp-1 protein levels in cultures transfected with either control<br />

or Insig-1 siRNA is shown in the right panel. Asterisks indicate precursor (*) <strong>and</strong> mature form (**) of<br />

Srebp-1.<br />

Fig. 6<br />

Induction of Insig1 by PB <strong>and</strong> AMPK in primary human hepatocytes. A, Cells from two different<br />

donors were incubated with 500µM PB <strong>and</strong> mRNA analyzed 50h thereafter. B, AMPK <strong>and</strong> PB<br />

synergistically activate human INSIG-1. Primary human hepatocytes from a third donor were infected<br />

with adenoviral particles encoding either control gene (B-Gal), a dominant negative form of AMPK<br />

(KD), the alpha-1 subunit of AMPK (AMPKα1) or the alpha-2 subunit of AMPK (AMPKα2).<br />

*p


MOL Manuscript #41012<br />

Table 1: Primers <strong>and</strong> Probes used for RT-qPCR<br />

Gene Forward Reverse Taqman-Probe<br />

Mouse:<br />

Gapdh CCAGAACATCATCCCTGCATC GGTCCTCAGTGTAGCCCAAGAT CCGCCTGGAGAAACCTGCCAAGTATG<br />

Cyp2b10 CAATGTTTAGTGGAGGAACTGCG CACTGGAAGAGGAACGTGGG CCCAGGGAGCCCCCCTGGA<br />

Cyp3a11 AGAACTTCTCCTTCCAGCCTTGTA GAGGGAGACTCATGCTCCAGTTA CTAAAGGTTGTGCCACGGGATGCAGT<br />

Srebp1c GGAGCCATGGATTGCACATT CCTGTCTCACCCCCAGCATA CAGCTCATCAACAACCAAGACAGTGACTTCC<br />

Scd-1* CCGGAGACCCTTAGATCGA TAGCCTGTAAAAGATTTCTGCAAACC<br />

Hmgcr* CGAGGAAAGACTGTGGTTTGTG CGTCAACCATAGCTTCCGTAGTT<br />

Insig-1* TGCAGATCCAGCGGAATGT CCAGGCGGAGGAGAAGATG<br />

Human:<br />

CYP2B6 ACATCGCCCTCCAGAGCTT GTCGGAAAATCTCTGAATCTCATAGA ACCGAGCCAAAATGCCATACACAGAGG<br />

CYP3A4 CATTCCTCATCCCAATTCTTGAAGT CCACTCGGTGCTTTTGTGTATCT CGAGGCGACTTTCTTTCATCCTTTTTACAGATTTTC<br />

INSIG-1* AGCCCCTACCCCAACACCT ACCACCCCAACCGAGAAGA<br />

SREBP1c* TCAGCGAGGCGGCTTTGGAGCAG CATGTCTTCGATGTCGGTCAG<br />

18s AGTCCCTGCCCTTTGTACACA CGATCCGAGGGCCTCACTA CGCCCGTCGCTACTACCGATTGG<br />

(*SYBR primers)<br />

25


mg/g Tissue<br />

Rel mRNA Level<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

Triglycerides<br />

* *<br />

Insig1<br />

**<br />

**<br />

** **<br />

Ctrl<br />

PB<br />

PCN<br />

TCPOBOP<br />

mg/g Tissue<br />

Rel mRNA Level<br />

Rel mRNA Level<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Cholesterol<br />

Srebp1c<br />

Scd1<br />

*<br />

*<br />

**<br />

*p < 0.05<br />

* * p < 0.005<br />

mg/dl<br />

Figure 1<br />

*<br />

*<br />

Serum Chemistry<br />

400<br />

TG<br />

350<br />

300<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

Rel mRNA Level<br />

Rel mRNA Level<br />

6 Srebp2<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

6 Hmgcr<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

*<br />

CHOL


kDa<br />

150<br />

100<br />

75<br />

50<br />

Rel. mRNA Level<br />

A<br />

B<br />

15<br />

12<br />

9<br />

6<br />

3<br />

1<br />

DMSO PB<br />

Cyp2b10<br />

Insig1<br />

*<br />

**<br />

kDa<br />

150<br />

100<br />

75<br />

50<br />

DMSO PCN TCPOBOP<br />

0 1 2 3 4 5<br />

Figure 2<br />

*<br />

**<br />

hrs after<br />

addition<br />

of drug


Relative mRNA Level<br />

Relative mRNA Level<br />

400 Cyp2b10<br />

350<br />

300<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

WT Double-KO<br />

**<br />

Relative mRNA Level<br />

- + - + PB - + - + PB<br />

Insig1 Triglycerides<br />

WT Double-KO<br />

20<br />

18<br />

WT Double-KO<br />

**<br />

16<br />

14<br />

mg/g Tissue<br />

3<br />

2.5 **<br />

Cyp3a11<br />

WT Double-KO<br />

1.5<br />

0.5<br />

0<br />

12<br />

10<br />

Figure 3<br />

2<br />

1<br />

8<br />

6<br />

4<br />

2<br />

0<br />

- + - + PB - + - + PB<br />

**


A<br />

RXR<br />

CAR<br />

PXR<br />

Anti-RXR<br />

kb from transcriptional start site<br />

7 6 5 4 3 2 1<br />

-3044<br />

-6252<br />

-5118<br />

-6252 -5881<br />

-6252 -5111<br />

-5871 -5111<br />

-5340 -5314<br />

-5340 -5314<br />

X<br />

DR4<br />

- + - - + +<br />

- - - ++ +-<br />

++<br />

B C<br />

+ - ++<br />

-<br />

-<br />

- - - - + - +<br />

**<br />

*<br />

Figure 4<br />

0<br />

vp16PXR<br />

vp16CAR<br />

0 1 2 3 4 5 6<br />

fold activation reporter fragment<br />

over empty reporter vector<br />

-5333<br />

-5319<br />

wt …CCTGAGGGTCAACAGAGGACACCTAG…<br />

mut …CCTGAGTTTAAACAGAGGACACCTAG…<br />

control<br />

CAR<br />

PXR<br />

Insig1-DR4<br />

No Ab<br />

Input<br />

GAPDH


A<br />

Fold induction reporter<br />

B<br />

Rel mRNA Level<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

Ctrl-siRNA<br />

mInsig1-DR4<br />

-<br />

+mPXR<br />

+mCAR<br />

**<br />

Insig1-siRNA<br />

-<br />

kDa<br />

150<br />

100<br />

75<br />

50<br />

Androstanol<br />

PCN<br />

Figure 5<br />

Androstanol/<br />

TCPOBOP<br />

*<br />

- + -<br />

**<br />

+ PB<br />

Ctrl-siRNA Insig1-siRNA


A<br />

Rel mRNA Level<br />

Rel mRNA Level<br />

4 INSIG1<br />

Donor 1 Donor 2<br />

3<br />

2<br />

1<br />

0<br />

16<br />

12<br />

8<br />

4<br />

0<br />

B<br />

Rel mRNA Level<br />

- + - + PB<br />

CYP2B6<br />

Donor 1 Donor 2<br />

- + - + PB<br />

14 INSIG1<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

*<br />

Rel mRNA Level<br />

Rel mRNA Level<br />

1.25 SREBP1c<br />

Donor 1 Donor 2<br />

1<br />

0.75<br />

0.5<br />

0.25<br />

400<br />

300<br />

200<br />

100<br />

**<br />

0<br />

CYP3A4<br />

Donor 1 Donor 2<br />

B-Gal KD AMPKalpha1 AMPKalpha2<br />

- - + - + - + PB<br />

Figure 6<br />

- + - + PB<br />

- + - + PB<br />

*<br />

**

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!